ArticlePDF Available

Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice

Rockefeller University Press
Journal of Experimental Medicine (JEM)
Authors:

Abstract and Figures

Interleukin 7 (IL-7) stimulates the proliferation of B cell progenitors, thymocytes, and mature T cells through an interaction with a high affinity receptor (IL-7R) belonging to the hematopoietin receptor superfamily. We have further addressed the role of IL-7 and its receptor during B and T cell development by generating mice genetically deficient in IL-7R. Mutant mice display a profound reduction in thymic and peripheral lymphoid cellularity. Analyses of lymphoid progenitor populations in IL-7R-deficient mice define precisely those developmental stages affected by the mutation and reveal a critical role for IL-7R during early lymphoid development. Significantly, these studies indicate that the phase of thymocyte expansion occurring before the onset of T cell receptor gene rearrangement is critically dependent upon, and mediated by the high affinity receptor for IL-7.
Content may be subject to copyright.
A preview of the PDF is not available
... Рецептор интерлейкина-7 и путь JAK/STAT Интерлейкин-7 (IL-7) и его рецептор необходимы для нормального созревания, функционирования и выживания Т-клеток [67]. Рецептор к IL-7 является гетеродимером и состоит из a-субъединицы, кодируемой геном IL7R, и общей g-цепи, кодируемой геном IL2RG. ...
Article
T-lymphoblastic lymphoma (T-LBL) is one of the most common non-Hodgkin lymphomas in children. According to the 2022 WHO classification, T-LBL and acute T-lymphoblastic leukemia are considered as a single disease since they both have T-cell precursors as a morphological substrate. In recent years, some progress has been made in the treatment of this disease, but the prognosis for relapses and refractory cases remains extremely unfavorable. One of the promising areas that can increase the effectiveness of therapy is the use of new treatment approaches that consider the molecular and biological features of this tumor. This review examines in detail the molecular aspects of the pathogenesis of T-LBL.
... 21 Similarly, mutation of the IL7R gene inhibits the development of human T cells, but in mice, it affects both T and B cell development. 22,23 Interspecies differences between humans and mice also result in different immune response processes. In humans, activation of Th2 cells results in the recruitment of eosinophils and the production of IgE by B cells, which is the main defense mechanism against schistosomiasis; however, mice use Th1 cell-produced IFNγ to combat parasite infection. ...
Article
Full-text available
The mouse genome has a high degree of homology with the human genome, and its physiological, biochemical, and developmental regulation mechanisms are similar to those of humans; therefore, mice are widely used as experimental animals. However, it is undeniable that interspecies differences between humans and mice can lead to experimental errors. The differences in the immune system have become an important factor limiting current immunological research. The application of immunodeficient mice provides a possible solution to these problems. By transplanting human immune cells or tissues, such as peripheral blood mononuclear cells or hematopoietic stem cells, into immunodeficient mice, a human immune system can be reconstituted in the mouse body, and the engrafted immune cells can elicit human‐specific immune responses. Researchers have been actively exploring the development and differentiation conditions of host recipient animals and grafts in order to achieve better immune reconstitution. Through genetic engineering methods, immunodeficient mice can be further modified to provide a favorable developmental and differentiation microenvironment for the grafts. From initially only being able to reconstruct single T lymphocyte lineages, it is now possible to reconstruct lymphoid and myeloid cells, providing important research tools for immunology‐related studies. In this review, we compare the differences in immune systems of humans and mice, describe the development history of human immune reconstitution from the perspectives of immunodeficient mice and grafts, and discuss the latest advances in enhancing the efficiency of human immune cell reconstitution, aiming to provide important references for immunological related researches.
... Our data show that IL7 in conjunction with RT can be used to control tumor growth in both HPV+ and HPV− preclinical models of HNSCC. IL7 is critical for lymphocyte development and maintenance, and knockout of the IL7R gene leads to a dearth of lymphocyte activity [47]. While IL7 has multiple roles along the T-cell developmental pathway, T-cell survival and maintenance in particular are regulated by IL7 secretion [21,24,48]. ...
Article
Full-text available
Clinically approved head and neck squamous cell carcinoma (HNSCC) immunotherapies manipulate the immune checkpoint blockade (ICB) axis but have had limited success outside of recurrent/metastatic disease. Interleukin-7 (IL7) has been shown to be essential for effector T-cell survival, activation, and proliferation. Here, we show that IL7 in combination with radiotherapy (RT) is effective in activating CD8 + T-cells for reducing tumor growth. Our studies were conducted using both human papillomavirus related and unrelated orthotopic HNSCC murine models. Immune populations from the tumor, draining lymph nodes, and blood were compared between treatment groups and controls using flow cytometry, proteomics, immunofluorescence staining, and RNA sequencing. Treatment with RT and IL7 (RT + IL7) resulted in significant tumor growth reduction, high CD8 T-cell tumor infiltration, and increased proliferation of T-cell progenitors in the bone marrow. IL7 also expanded a memory-like subpopulation of CD8 T-cells. These results indicate that IL7 in combination with RT can serve as an effective immunotherapy strategy outside of the conventional ICB axis to drive the antitumor activity of CD8 T-cells.
... Hematopoietic stem cells will preferably differentiate to become the earliest multilymphoid progenitors upon stimulation by interleukin (IL)-7 (31). From that point, common lymphoid progenitor cells can continue dividing and eventually differentiate themselves to become thymocytes ("pre-T" cells), pre-B cells, or pre-NK cells, dedicated progenitors of the three main types of lymphocytes. ...
Article
Full-text available
Ionising radiations can have a wide range of impacts on tumour-immune interactions, which are being studied with the greatest interest and at an accelerating pace by the medical community. Despite its undeniable immunostimulatory potential, it clearly appears that radiotherapy as it is prescribed and delivered nowadays often alters the host’s immunity towards a suboptimal state. This may impair the full recovery of a sustained and efficient anti-tumour immunosurveillance post-treatment. An emerging concept is arising from this awareness and consists in re-considering the way of designing radiation treatment plannings, notably by taking into account the individualised risks of deleterious radio-induced immune alteration that can be deciphered from the planned beam trajectory through lymphocyte-rich organs. In this review, we critically appraise key aspects to consider while planning immunologically-fitted radiotherapy, including the challenges linked to the identification of new dose constraints to immune-rich structures. We also discuss how pharmacological immunomodulation could be advantageously used in combination with radiotherapy to compensate for the radio-induced loss, for example with (i) agonists of interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15 or IL-21, similarly to G-CSF being used for the prophylaxis of severe chemo-induced neutropenia, or with (ii) myeloid-derived suppressive cells (MDSC) blockers.
... DN thymocytes express large amounts of the IL-7 receptor, rendering them responsive to intrathymic IL-7, which is produced by thymic epithelial cells and dendritic cells (DCs) at the cortico-medullary junction (12). In this regard, genetic deficiencies in IL-7 or IL-7 receptor manifest in severely impaired thymopoiesis and dramatically decreased thymic cellularity, because the lack of IL-7 signaling in DN thymocytes impairs their proliferation and expansion (13,14). Interestingly, different levels of IL-7 receptor expression among thymic DN cells were proposed being associated with γδ vs. αβ T lineage differentiation, suggesting an instructive role for IL-7 in early T cell lineage decision (15). ...
Article
Full-text available
The cytokine IL-7 plays critical and nonredundant roles in T cell immunity so that the abundance and availability of IL-7 act as key regulatory mechanisms in T cell immunity. Importantly, IL-7 is not produced by T cells themselves but primarily by non-lymphoid lineage stromal cells and epithelial cells that are limited in their numbers. Thus, T cells depend on cell extrinsic IL-7, and the amount of in vivo IL-7 is considered a major factor in maximizing and maintaining the number of T cells in peripheral tissues. Moreover, IL-7 provides metabolic cues and promotes the survival of both naïve and memory T cells. Thus, IL-7 is also essential for the functional fitness of T cells. In this regard, there has been an extensive effort trying to increase the protein abundance of IL-7 in vivo, with the aim to augment T cell immunity and harness T cell functions in anti-tumor responses. Such approaches started under experimental animal models, but they recently culminated into clinical studies, with striking effects in re-establishing T cell immunity in immunocompromised patients, as well as boosting anti-tumor effects. Depending on the design, glycosylation, and the structure of recombinantly engineered IL-7 proteins and their mimetics, recombinant IL-7 molecules have shown dramatic differences in their stability, efficacy, cellular effects, and overall immune functions. The current review is aimed to summarize the past and present efforts in the field that led to clinical trials, and to highlight the therapeutical significance of IL-7 biology as a master regulator of T cell immunity.
... Furthermore, patients harboring LOF mutations in the IL-7 receptor alpha (IL-7Rα) chain display a T-B+NK+ SCID phenotype [98,99]. Mice with IL-7Rα deficiency again possessed a more severe SCID that included B cell involvement [100,101]. Zebrafish carrying an inactivating mutation of Il-7rα exhibited reduced T cells, but unaffected B cells, again similar to humans, with NK cells not characterized in this study [89]. STAT5B-deficient humans also displayed immunodeficiency and growth defects [102], which closely resembled those observed in Zebrafish Stat5.1 LOF mutants [103]. ...
Article
Full-text available
The Janus kinase (JAK) family is a small group of protein tyrosine kinases that represent a central component of intracellular signaling downstream from a myriad of cytokine receptors. The JAK3 family member performs a particularly important role in facilitating signal transduction for a key set of cytokine receptors that are essential for immune cell development and function. Mutations that impact JAK3 activity have been identified in a number of human diseases, including somatic gain-of-function (GOF) mutations associated with immune cell malignancies and germline loss-of-function (LOF) mutations associated with immunodeficiency. The structure, function and impacts of both GOF and LOF mutations of JAK3 are highly conserved, making animal models highly informative. This review details the biology of JAK3 and the impact of its perturbation in immune cell-related diseases, including relevant animal studies.
Article
The size and condition of the peripheral CD4 T cell population determine the capacity of the immune response. Under homeostatic conditions, the size of the peripheral CD4 T cell population is maintained through turnover and survival. However, the underlying mechanisms remain inadequately understood. Here, we observed a significant decrease in the percentage of CD4 T cells in the periphery following the targeted deletion of the Paxbp1 gene in mouse T cells. In the absence of Paxbp1, naïve CD4 T cells displayed reduced surface interleukin-7 receptor levels and a decreased capacity to respond to survival signals mediated by interleukin-7. In addition, naïve CD4 T cells deficient in Paxbp1 demonstrated impaired T cell antigen receptor signalling, compromised cell cycle entry, decreased proliferation, and increased apoptosis following stimulation, all of which contributed to the reduction in the number of peripheral CD4 T cells. Therefore, our study highlights the indispensable role of Paxbp1 in maintaining peripheral CD4 T cell homeostasis.
Article
Naturally occurring gain-of-function (GOF) mutants have been identified in patients for a variety of cytokine receptors. Although this constitutive activation of cytokine receptors is strongly associated with malignant disorders, ligand-independent receptor activation is also a useful tool in synthetic biology e.g. to improve adoptive cellular therapies with genetically modified T-cells. Balanced Interleukin (IL-)7 signaling via a heterodimer of IL-7 receptor (IL-7Rα) and the common γ-chain (γc) controls T- and B-cell development and expansion, whereas uncontrolled IL-7 signaling can drive acute lymphoid leukemia (ALL) development. The ALL-driver mutation PPCL in the transmembrane domain of IL-7Rα is a mutational insertion of the four amino acids proline-proline-cysteine-leucine and leads to ligand-independent receptor dimerization and constitutive activation. We showed here in the cytokine-dependent pre-B-cell line Ba/F3 that the PPCL-insertion in a synthetic version of the IL-7Rα induced γc-independent STAT5 and ERK phosphorylation and also proliferation of the cells and that booster-stimulation by arteficial ligands additionally generated non-canonical STAT3 phosphorylation via the synthetic IL-7Rα-PPCL-receptors. Transfer of the IL-7Rα transmembrane domain with the PPCL insertion into natural and synthetic cytokine receptor chains of the IL-6, IL-12 and Interferon families also resulted in constitutive receptor signaling. In conclusion, our data suggested that the insertion of the mutated PPCL IL-7Rα transmembrane domain is an universal approach to generate ligand-independent, constitutively active cytokine receptors.
Article
Full-text available
Rheumatoid arthritis (RA) is an ongoing inflammatory condition that affects the joints and can lead to severe damage to cartilage and bones, resulting in significant disability. This condition occurs when the immune system becomes overactive, causing osteoclasts, cells responsible for breaking down bone, to become more active than necessary, leading to bone breakdown. RA disrupts the equilibrium between osteoclasts and osteoblasts, resulting in serious complications such as localized bone erosion, weakened bones surrounding the joints, and even widespread osteoporosis. Antibodies against the receptor activator of nuclear factor-κB ligand (RANKL), a crucial stimulator of osteoclast differentiation, have shown great effectiveness both in laboratory settings and actual patient cases. Researchers are increasingly focusing on osteoclasts as significant contributors to bone erosion in RA. Given that RA involves an overactive immune system, T cells and B cells play a pivotal role by intensifying the immune response. The imbalance between Th17 cells and Treg cells, premature aging of T cells, and excessive production of antibodies by B cells not only exacerbate inflammation but also accelerate bone destruction. Understanding the connection between the immune system and osteoclasts is crucial for comprehending the impact of RA on bone health. By delving into the immune mechanisms that lead to joint damage, exploring the interactions between the immune system and osteoclasts, and investigating new biomarkers for RA, we can significantly improve early diagnosis, treatment, and prognosis of this condition.
Article
Full-text available
Analysis of mice carrying mutant T-cell antigen receptor (TCR) genes indicates that TCR-beta gene rearrangement or expression is critical for the differentiation of CD4-CD8- thymocytes to CD4+CD8+ thymocytes, as well as for the expansion of the pool of CD4+CD8+ cells. TCR-alpha is irrelevant in these developmental processes. The development of gamma delta T cells does not depend on either TCR-alpha or TCR-beta.
Article
Full-text available
The protein Lck (p56lck) has a relative molecular mass of 56,000 and belongs to the Src family of tyrosine kinases. It is expressed exclusively in lymphoid cells, predominantly in thymocytes and peripheral T cells. Lck associates specifically with the cytoplasmic domains of both CD4 and CD8 T-cell surface glycoproteins and interacts with the beta-chain of the interleukin-2 receptor, which implicates Lck activity in signal transduction during thymocyte ontogeny and activation of mature T cells. Here we generate an lck null mutation by homologous recombination in embryonic stem cells to evaluate the role of p56lck in T-cell development and activation. Lck-deficient mice show a pronounced thymic atrophy, with a dramatic reduction in the double-positive (CD4+CD8+) thymocyte population. Mature, single-positive thymocytes are not detectable in these mice and there are only very few peripheral T cells. These results illustrate the crucial role of this T-cell-specific tyrosine kinase in the thymocyte development.
Article
Analysis of mice carrying mutant T-cell antigen receptor (TCR) genes indicates that TCR-β gene rearrangement or expression is critical for the differentiation of CD4-CD8- thymocytes to CD4+CD8+ thymocytes, as well as for the expansion of the pool of CD4+CD8+ cells. TCR-α is irrelevant in these developmental processes. The development of γδ T cells does not depend on either TCR-α or TCR-β.
Article
The role of IL-7 in the stromal cell-dependent B cell development was investigated using two stromal cell clones, ST2 and PA6; the former supports B lymphopoiesis while the latter can not. We demonstrate here that: (a) the ability of the stromal cell clone to produce IL-7 correlates well with the stromal cell activity to support B lymphopoiesis; (b) IL-7 production by ST2 is inducible rather than constitutive; (c) the IL-7-dependent B cell itself is a potent inducer of IL-7 production by ST2; (d) addition of rIL-7 to the PA6 layer renders this in vitro environment B lymphopoietic; and (e) the differentiation from early B progenitor to pre-B cell requires both IL-7 and other stromal cell molecule(s) yet to be identified.
Article
The effects of interleukin 7 (IL-7) on the growth and differentiation of murine B cell progenitors has been well characterized using in vitro culture methods. We have investigated the role of IL-7 in vivo using a monoclonal antibody that neutralizes IL-7. We find that treatment of mice with this antibody completely inhibits the development of B cell progenitors from the pro-B cell stage forward. We also provide evidence that all peripheral B cells, including those of the B-1 and conventional lineages, are derived from IL-7-dependent precursors. The results are consistent with the rapid turnover of B cell progenitors in the marrow, but a slow turnover of mature B cells in the periphery. In addition to effects on B cell development, anti-IL-7 treatment substantially reduced thymus cellularity, affecting all major thymic subpopulations.
Article
The gamma chain of the interleukin-2 (IL-2) receptor is shared with the functional IL-4 receptor and is causatively related to X-linked severe combined immunodeficiency (XSCID), which is ascribed to a profound T cell defect. Studies with monoclonal antibodies specific for the IL-2 receptor gamma chain showed that the gamma chain participates in the functional high-affinity receptor complexes for IL-7 that are involved in the differentiation of T and B cells. Participation of the gamma subunit in more than one receptor may enable the elucidation of the mechanisms of XSCID development and lymphocyte differentiation.
Article
Studies from several laboratories have provided evidence that distinct stromal cell-derived signals are involved in the maturation of pre-B cells into surface Ig expressing B lymphocytes. In order to define the stage of development at which these stimuli act, various polymerase chain reaction strategies were used to characterize the status of kappa L chain gene rearrangements in nontransformed, stromal cell dependent pre-B cells. These cells were obtained from lymphoid colonies whose growth was potentiated by factors from a stromal cell line. kappa L chain genes in cells from many of these colonies were rearranged, and analysis of the Jk genes used indicated a bias toward the most 3' loci. However, the use of a reverse transcriptase PCR strategy failed to detect mature kappa transcripts, indicating that stromal cell mediators exist that allow pre-B cells to progress to the stage at which L chain genes are rearranged but not expressed. Reverse transcriptase PCR further revealed that no transcripts for c-kit (the receptor for kit-ligand) and the IL-7R could be detected in these cells. This suggests that these receptors are no longer expressed by the time cells have undergone kappa rearrangements and minimize a role for stromal cell-derived kit-ligand and IL-7 in mediating the pre-B to B cell transition.
Article
IL-7 has previously been shown to stimulate proliferation of immature CD4-CD8- thymocytes from fetal and adult mice. We show that IL-7 mRNA levels are much higher in the day-14 fetal thymus than in the adult thymus, i.e., when the thymus is primarily composed of CD4-CD8- cells. Inasmuch as IL-7 may, therefore, be one of the first cytokines to which pre-T cells from the fetal liver are exposed when they migrate to the thymus during development, we hypothesized that IL-7 may promote differentiation of pre-T cells from the fetal liver. We found that human rIL-7 promotes growth of cells obtained from the liver of fetal mice at day 14 of gestation. Moreover, we found that IL-7 stimulated expression of mRNA encoding TCR-gamma, alpha-, and beta-genes. The size of the transcripts induced by IL-7 suggests that TCR-gamma was expressed in a rearranged form in response to IL-7, whereas TCR-alpha and -beta genes appeared to be expressed from nonrearranged loci. Culture of fetal liver cells for 7 days with IL-7 also caused the appearance of a population of cells expressing Thy-1 and Pgp-1 Ag, a phenotype similar to that seen in the early (day-14) fetal thymus. The Thy-1+ Pgp-1+ population was enriched in TCR-gamma mRNA, as determined by flow cytometric sorting followed by Northern blot analysis. These data support the hypothesis that IL-7 can stimulate some of the early events associated with thymocyte differentiation in the fetus.
Article
This chapter provides an overview of the properties of mature thymus-derived (T) cells, including both the molecules they use for recognition and the molecules they use in responses. The diversity of characteristics that are loosely considered “T cell specific” allows tackling critically the issue of the way they are coordinated in development. T lymphocytes play central roles in immune responses and in the regulation of cell growth for several hematopoietic lineages. The importance of these cells is derived from their unique integration of specific antigen recognition with an impressively versatile, highly regulated array of possible effector functions. T cell development is divided into four phases. First is an expansion phase prior to T cell receptor (TCR) rearrangement, which appears to be correlated with programming of at least some response genes for inducibility. Second is a controlled phase of TCR gene rearrangement. Third is the complex process of selection. Whereas peripheral T cells can undergo forms of positive selection and negative selection, neither is exactly the same phenomenon that occurs in the thymic cortex. The fourth stage is the reconversion of the positively selected TCR αβ cell into a responsive effector cell.